Your browser doesn't support javascript.
loading
Mostrar: 20 | 50 | 100
Resultados 1 - 20 de 25
Filtrar
1.
Cell Rep ; 40(9): 111267, 2022 08 30.
Artículo en Inglés | MEDLINE | ID: mdl-36044855

RESUMEN

Rhabdomyosarcoma (RMS) is the most common soft-tissue sarcoma of childhood characterized by the inability to exit the proliferative myoblast-like stage. The alveolar fusion positive subtype (FP-RMS) is the most aggressive and is mainly caused by the expression of PAX3/7-FOXO1 oncoproteins, which are challenging pharmacological targets. Here, we show that the DEAD box RNA helicase 5 (DDX5) is overexpressed in alveolar RMS cells and that its depletion and pharmacological inhibition decrease FP-RMS viability and slow tumor growth in xenograft models. Mechanistically, we provide evidence that DDX5 functions upstream of the EHMT2/AKT survival signaling pathway, by directly interacting with EHMT2 mRNA, modulating its stability and consequent protein expression. We show that EHMT2 in turns regulates PAX3-FOXO1 activity in a methylation-dependent manner, thus sustaining FP-RMS myoblastic state. Together, our findings identify another survival-promoting loop in FP-RMS and highlight DDX5 as a potential therapeutic target to arrest RMS growth.


Asunto(s)
ARN Helicasas DEAD-box , Rabdomiosarcoma Alveolar , Rabdomiosarcoma Embrionario , Rabdomiosarcoma , Línea Celular Tumoral , ARN Helicasas DEAD-box/genética , ARN Helicasas DEAD-box/metabolismo , Regulación Neoplásica de la Expresión Génica , Antígenos de Histocompatibilidad , N-Metiltransferasa de Histona-Lisina/metabolismo , Humanos , Proteínas de Fusión Oncogénica/metabolismo , Factores de Transcripción Paired Box/genética , ARN Helicasas/metabolismo , Rabdomiosarcoma/metabolismo , Rabdomiosarcoma Alveolar/genética , Rabdomiosarcoma Alveolar/metabolismo , Rabdomiosarcoma Alveolar/patología
2.
Cells ; 11(7)2022 03 30.
Artículo en Inglés | MEDLINE | ID: mdl-35406733

RESUMEN

Mitochondrial bioenergetics are progressively acquiring significant pathophysiological roles. Specifically, mitochondria in general and Electron Respiratory Chain in particular are gaining importance as unintentional targets of different drugs. The so-called PPAR ligands are a class of drugs which not only link and activate Peroxisome Proliferator-Activated Receptors but also show a myriad of extrareceptorial activities as well. In particular, they were shown to inhibit NADH coenzyme Q reductase. However, the molecular picture of this intriguing bioenergetic derangement has not yet been well defined. Using high resolution respirometry, both in permeabilized and intact HepG2 cells, and a proteomic approach, the mitochondrial bioenergetic damage induced by various PPAR ligands was evaluated. Results show a derangement of mitochondrial oxidative metabolism more complex than one related to a simple perturbation of complex I. In fact, a partial inhibition of mitochondrial NADH oxidation seems to be associated not only with hampered ATP synthesis but also with a significant reduction in respiratory control ratio, spare respiratory capacity, coupling efficiency and, last but not least, serious oxidative stress and structural damage to mitochondria.


Asunto(s)
Receptores Activados del Proliferador del Peroxisoma , Proteómica , Complejo I de Transporte de Electrón/metabolismo , Hipoglucemiantes , Ligandos , Mitocondrias/metabolismo , Receptores Activados del Proliferador del Peroxisoma/metabolismo
3.
Cells ; 11(6)2022 03 20.
Artículo en Inglés | MEDLINE | ID: mdl-35326503

RESUMEN

RNA-binding proteins (RBPs) play important roles in modulating miRNA-mediated mRNA target repression. Argonaute2 (Ago2) is an essential component of the RNA-induced silencing complex (RISC) that plays a central role in silencing mechanisms via small non-coding RNA molecules known as siRNAs and miRNAs. Small RNAs loaded into Argonaute proteins catalyze endoribonucleolytic cleavage of target RNAs or recruit factors responsible for translational silencing and mRNA target destabilization. In previous studies we have shown that KCC2, a neuronal Cl (-) extruding K (+) Cl (-) co-transporter 2, is regulated by miR-92 in neuronal cells. Searching for Ago2 partners by immunoprecipitation and LC-MS/MS analysis, we isolated among other proteins the Serpine mRNA binding protein 1 (SERBP1) from SH-SY5Y neuroblastoma cells. Exploring the role of SERBP1 in miRNA-mediated gene silencing in SH-SY5Y cells and primary hippocampal neurons, we demonstrated that SERBP1 silencing regulates KCC2 expression through the 3' untranslated region (UTR). In addition, we found that SERBP1 as well as Ago2/miR-92 complex bind to KCC2 3'UTR. Finally, we demonstrated the attenuation of miR-92-mediated repression of KCC2 3'UTR by SERBP1 silencing. These findings advance our knowledge regarding the miR-92-mediated modulation of KCC2 translation in neuronal cells and highlight SERBP1 as a key component of this gene regulation.


Asunto(s)
MicroARNs , Simportadores , Regiones no Traducidas 3' , Cromatografía Liquida , MicroARNs/genética , MicroARNs/metabolismo , Neuronas/metabolismo , ARN Mensajero/genética , Complejo Silenciador Inducido por ARN/genética , Simportadores/genética , Espectrometría de Masas en Tándem
4.
Open Biol ; 11(12): 210276, 2021 12.
Artículo en Inglés | MEDLINE | ID: mdl-34847775

RESUMEN

Amplification of the proto-oncogene MYCN is a key molecular aberration in high-risk neuroblastoma and predictive of poor outcome in this childhood malignancy. We investigated the role of MYCN in regulating the protein cargo of extracellular vesicles (EVs) secreted by tumour cells that can be internalized by recipient cells with functional consequences. Using a switchable MYCN system coupled to mass spectrometry analysis, we found that MYCN regulates distinct sets of proteins in the EVs secreted by neuroblastoma cells. EVs produced by MYCN-expressing cells or isolated from neuroblastoma patients induced the Warburg effect, proliferation and c-MYC expression in target cells. Mechanistically, we linked the cancer-promoting activity of EVs to the glycolytic kinase pyruvate kinase M2 (PKM2) that was enriched in EVs secreted by MYC-expressing neuroblastoma cells. Importantly, the glycolytic enzymes PKM2 and hexokinase II were detected in the EVs circulating in the bloodstream of neuroblastoma patients, but not in those of non-cancer children. We conclude that MYC-activated cancers might spread oncogenic signals to remote body locations through EVs.


Asunto(s)
Proteínas Portadoras/metabolismo , Vesículas Extracelulares/enzimología , Hexoquinasa/metabolismo , Proteínas de la Membrana/metabolismo , Proteína Proto-Oncogénica N-Myc/genética , Neuroblastoma/genética , Proteómica/métodos , Hormonas Tiroideas/metabolismo , Proteínas Portadoras/sangre , Línea Celular Tumoral , Proliferación Celular , Niño , Amplificación de Genes , Regulación Neoplásica de la Expresión Génica , Redes Reguladoras de Genes , Glucólisis , Hexoquinasa/sangre , Humanos , Espectrometría de Masas , Proteínas de la Membrana/sangre , Neuroblastoma/sangre , Fosforilación , Hormonas Tiroideas/sangre , Proteínas de Unión a Hormona Tiroide
5.
Cell Death Dis ; 12(6): 558, 2021 05 29.
Artículo en Inglés | MEDLINE | ID: mdl-34052831

RESUMEN

Epithelial ovarian cancer (EOC) is a highly heterogeneous disease with a high death rate mainly due to the metastatic spread. The expression of MDM4, a well-known p53-inhibitor, is positively associated with chemotherapy response and overall survival (OS) in EOC. However, the basis of this association remains elusive. We show that in vivo MDM4 reduces intraperitoneal dissemination of EOC cells, independently of p53 and an immune-competent background. By 2D and 3D assays, MDM4 impairs the early steps of the metastatic process. A 3D-bioprinting system, ad hoc developed by co-culturing EOC spheroids and endothelial cells, showed reduced dissemination and intravasation into vessel-like structures of MDM4-expressing cells. Consistent with these data, high MDM4 levels protect mice from ovarian cancer-related death and, importantly, correlate with increased 15 y OS probability in large data set analysis of 1656 patients. Proteomic analysis of EOC 3D-spheroids revealed decreased protein synthesis and mTOR signaling, upon MDM4 expression. Accordingly, MDM4 does not further inhibit cell migration when its activity towards mTOR is blocked by genetic or pharmacological approaches. Importantly, high levels of MDM4 reduced the efficacy of mTOR inhibitors in constraining cell migration. Overall, these data demonstrate that MDM4 impairs EOC metastatic process by inhibiting mTOR activity and suggest the usefulness of MDM4 assessment for the tailored application of mTOR-targeted therapy.


Asunto(s)
Proteínas de Ciclo Celular/metabolismo , Neoplasias Ováricas/genética , Proteómica/métodos , Proteínas Proto-Oncogénicas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Femenino , Humanos , Ratones , Metástasis de la Neoplasia , Neoplasias Ováricas/mortalidad , Análisis de Supervivencia
6.
Cancer Genomics Proteomics ; 17(3): 225-236, 2020.
Artículo en Inglés | MEDLINE | ID: mdl-32345664

RESUMEN

BACKGROUND: Malignant pleural mesothelioma (MPM) a rare neoplasm linked to asbestos exposure is characterized by a poor prognosis. Soluble mesothelin is currently considered the most specific diagnostic biomarker. The aim of the study was to identify novel biomarkers by proteomic analysis of two MPM cell lines secretome. MATERIALS AND METHODS: The protein patterns of MPM cells secretome were examined and compared to a non-malignant mesothelial cell line using two-dimensional gel electrophoresis coupled to mass spectrometry. Serum levels of candidate biomarkers were determined in MPM patients and control subjects. RESULTS: Two up-regulated proteins involved in cancer biology, prosaposin and quiescin Q6 sulfhydryl oxidase 1, were considered candidate biomarkers. Serum levels of both proteins were significantly higher in MPM patients than control subjects. Combining the data of each receiver-operating characteristic analysis predicted a good diagnostic accuracy. CONCLUSION: A panel of the putative biomarkers represents a promising tool for MPM diagnosis.


Asunto(s)
Biomarcadores de Tumor/sangre , Neoplasias Pulmonares/sangre , Mesotelioma/sangre , Neoplasias Pleurales/sangre , Proteoma/metabolismo , Anciano , Estudios de Casos y Controles , Línea Celular Tumoral , Femenino , Proteínas Ligadas a GPI/sangre , Humanos , Neoplasias Pulmonares/patología , Masculino , Mesotelina , Mesotelioma/patología , Mesotelioma Maligno , Persona de Mediana Edad , Oxidorreductasas actuantes sobre Donantes de Grupos Sulfuro/sangre , Neoplasias Pleurales/patología , Curva ROC , Saposinas/sangre , Vías Secretoras
7.
Int J Mol Sci ; 21(1)2019 Dec 21.
Artículo en Inglés | MEDLINE | ID: mdl-31877774

RESUMEN

In order to effectively develop personalized medicine for kidney diseases we urgently need to develop highly accurate biomarkers for use in the clinic, since current biomarkers of kidney damage (changes in serum creatinine and/or urine albumin excretion) apply to a later stage of disease, lack accuracy, and are not connected with molecular pathophysiology. Analysis of urine peptide content (urinary peptidomics) has emerged as one of the most attractive areas in disease biomarker discovery. Urinary peptidome analysis allows the detection of short and long-term physiological or pathological changes occurring within the kidney. Urinary peptidomics has been applied extensively for several years now in renal patients, and may greatly improve kidney disease management by supporting earlier and more accurate detection, prognostic assessment, and prediction of response to treatment. It also promises better understanding of kidney disease pathophysiology, and has been proposed as a "liquid biopsy" to discriminate various types of renal disorders. Furthermore, proteins being the major drug targets, peptidome analysis may allow one to evaluate the effects of therapies at the protein signaling pathway level. We here review the most recent findings on urinary peptidomics in the setting of the most common kidney diseases.


Asunto(s)
Enfermedades Renales/orina , Péptidos/orina , Proteómica/métodos , Biomarcadores/química , Biomarcadores/orina , Humanos , Enfermedades Renales/patología , Espectrometría de Masas/métodos , Péptidos/química , Medicina de Precisión/métodos , Urinálisis/métodos
8.
Int J Mol Sci ; 20(10)2019 May 24.
Artículo en Inglés | MEDLINE | ID: mdl-31137614

RESUMEN

A number of factors can trigger amyotrophic lateral sclerosis (ALS), although its precise pathogenesis is still uncertain. In a previous study done by us, poisonous liquoral levels of hydrogen sulphide (H2S) in sporadic ALS patients were reported. In the same study very high concentrations of H2S in the cerebral tissues of the familial ALS (fALS) model of the SOD1G93A mouse, were measured. The objective of this study was to test whether decreasing the levels of H2S in the fALS mouse could be beneficial. Amino-oxyacetic acid (AOA)-a systemic dual inhibitor of cystathionine-ß-synthase and cystathionine-γ lyase (two key enzymes in the production of H2S)-was administered to fALS mice. AOA treatment decreased the content of H2S in the cerebral tissues, and the lifespan of female mice increased by approximately ten days, while disease progression in male mice was not affected. The histological evaluation of the spinal cord of the females revealed a significant increase in GFAP positivity and a significant decrease in IBA1 positivity. In conclusion, the results of the study indicate that, in the animal model, the inhibition of H2S production is more effective in females. The findings reinforce the need to adequately consider sex as a relevant factor in ALS.


Asunto(s)
Ácido Aminooxiacético/farmacología , Esclerosis Amiotrófica Lateral/metabolismo , Cistationina betasintasa/antagonistas & inhibidores , Inhibidores Enzimáticos/farmacología , Sulfuro de Hidrógeno/metabolismo , Ácido Aminooxiacético/uso terapéutico , Esclerosis Amiotrófica Lateral/tratamiento farmacológico , Esclerosis Amiotrófica Lateral/genética , Animales , Encéfalo/efectos de los fármacos , Encéfalo/metabolismo , Células Cultivadas , Inhibidores Enzimáticos/uso terapéutico , Femenino , Masculino , Ratones , Ratones Endogámicos C57BL , Neuroglía/efectos de los fármacos , Factores Sexuales , Superóxido Dismutasa-1/genética
9.
J Proteome Res ; 17(12): 4307-4314, 2018 12 07.
Artículo en Inglés | MEDLINE | ID: mdl-30284448

RESUMEN

Mitochondria are undeniably the cell powerhouse, directly affecting cell survival and fate. Growing evidence suggest that mitochondrial protein repertoire affects metabolic activity and plays an important role in determining cell proliferation/differentiation or quiescence shift. Consequently, the bioenergetic status of a cell is associated with the quality and abundance of the mitochondrial populations and proteomes. Mitochondrial morphology changes in the development of different cellular functions associated with metabolic switches. It is therefore reasonable to speculate that different cell lines do contain different mitochondrial-associated proteins, and the investigation of these pools may well represent a source for mining missing proteins (MPs). A very effective approach to increase the number of IDs through mass spectrometry consists of reducing the complexity of the biological samples by fractionation. The present study aims at investigating the mitochondrial proteome of five phenotypically different cell lines, possibly expressing some of the MPs, through an enrichment-fractionation approach at the organelle and protein level. We demonstrate a substantial increase in the proteome coverage, which, in turn, increases the likelihood of detecting low abundant proteins, often falling in the category of MPs, and resulting, for the present study, in the identification of METTL12, FAM163A, and RGS13. All MS data have been deposited to the MassIVE data repository ( https://massive.ucsd.edu ) with the data set identifier MSV000082409 and PXD010446.


Asunto(s)
Mitocondrias/química , Proteínas Mitocondriales/análisis , Proteoma/análisis , Línea Celular , Fraccionamiento Químico , Bases de Datos de Proteínas , Humanos , Espectrometría de Masas/métodos , Proteínas de la Membrana/análisis , Metiltransferasas/análisis , Proteínas de Neoplasias/análisis , Proteómica/métodos , Proteínas RGS/análisis
10.
Expert Rev Proteomics ; 15(8): 683-696, 2018 08.
Artículo en Inglés | MEDLINE | ID: mdl-30058389

RESUMEN

INTRODUCTION: The development of precision medicine requires advanced technologies to address the multifactorial disease stratification and to support personalized treatments. Among omics techniques, proteomics based on Mass Spectrometry (MS) is becoming increasingly relevant in clinical practice allowing a phenotypic characterization of the dynamic functional status of the organism. From this perspective, Matrix Assisted Laser Desorption Ionization Time of Flight (MALDI-TOF) MS is a suitable platform for providing a high-throughput support to clinics. Areas covered: This review aims to provide an updated overview of MALDI-TOF MS applications in clinical proteomics. The most relevant features of this analysis have been discussed, highlighting both pre-analytical and analytical factors that are crucial in proteomics studies. Particular emphasis is placed on biofluids proteomics for biomarkers discovery and on recent progresses in clinical microbiology, drug monitoring, and minimal residual disease (MRD). Expert commentary: Despite some analytical limitations, the latest technological advances together with the easiness of use, the low time and low cost consuming and the high throughput are making MALDI-TOF MS instruments very attractive for the clinical practice. These features offer a significant potential for the routine of the clinical laboratory and ultimately for personalized medicine.


Asunto(s)
Proteómica , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción/métodos , Líquidos Corporales/metabolismo , Descubrimiento de Drogas , Humanos , Neoplasia Residual/metabolismo
11.
Ther Clin Risk Manag ; 14: 1-9, 2018.
Artículo en Inglés | MEDLINE | ID: mdl-29296087

RESUMEN

The success and the quality of hemodialysis therapy are mainly related to both clearance and biocompatibility properties of the artificial membrane packed in the hemodialyzer. Performance of a membrane is strongly influenced by its interaction with the plasma protein repertoire during the extracorporeal procedure. Recognition that a number of medium-high molecular weight solutes, including proteins and protein-bound molecules, are potentially toxic has prompted the development of more permeable membranes. Such membrane engineering, however, may cause loss of vital proteins, with membrane removal being nonspecific. In addition, plasma proteins can be adsorbed onto the membrane surface upon blood contact during dialysis. Adsorption can contribute to the removal of toxic compounds and governs the biocompatibility of a membrane, since surface-adsorbed proteins may trigger a variety of biologic blood pathways with pathophysiologic consequences. Over the last years, use of proteomic approaches has allowed polypeptide spectrum involved in the process of hemodialysis, a key issue previously hampered by lack of suitable technology, to be assessed in an unbiased manner and in its full complexity. Proteomics has been successfully applied to identify and quantify proteins in complex mixtures such as dialysis outflow fluid and fluid desorbed from dialysis membrane containing adsorbed proteins. The identified proteins can also be characterized by their involvement in metabolic and signaling pathways, molecular networks, and biologic processes through application of bioinformatics tools. Proteomics may thus provide an actual functional definition as to the effect of a membrane material on plasma proteins during hemodialysis. Here, we review the results of proteomic studies on the performance of hemodialysis membranes, as evaluated in terms of solute removal efficiency and blood-membrane interactions. The evidence collected indicates that the information provided by proteomic investigations yields improved molecular and functional knowledge and may lead to the development of more efficient membranes for the potential benefit of the patient.

12.
Mol Cancer ; 16(1): 55, 2017 03 07.
Artículo en Inglés | MEDLINE | ID: mdl-28270148

RESUMEN

BACKGROUND: Many tumor-related factors have shown the ability to affect metabolic pathways by paving the way for cancer-specific metabolic features. Here, we investigate the regulation of mTORC1 by MDM4, a p53-inhibitor with oncogenic or anti-survival activities depending on cell growth conditions. METHOD: MDM4-mTOR relationship was analysed through experiments of overexpression or silencing of endogenous proteins in cell culture and using purified proteins in vitro. Data were further confirmed in vivo using a transgenic mouse model overexpressing MDM4. Additionally, the Cancer Genome Atlas (TCGA) database (N = 356) was adopted to analyze the correlation between MDM4 and mTOR levels and 3D cultures were used to analyse the p53-independent activity of MDM4. RESULTS: Following nutrient deprivation, MDM4 impairs mTORC1 activity by binding and inhibiting the kinase mTOR, and contributing to maintain the cytosolic inactive pool of mTORC1. This function is independent of p53. Inhibition of mTORC1 by MDM4 results in reduced phosphorylation of the mTOR downstream target p70S6K1 both in vitro and in vivo in a MDM4-transgenic mouse. Consistently, MDM4 reduces cell size and proliferation, two features controlled by p70S6K1, and, importantly, inhibits mTORC1-mediated mammosphere formation. Noteworthy, MDM4 transcript levels are significantly reduced in breast tumors characterized by high mTOR levels. CONCLUSION: Overall, these data identify MDM4 as a nutrient-sensor able to inhibit mTORC1 and highlight its metabolism-related tumor-suppressing function.


Asunto(s)
Complejos Multiproteicos/metabolismo , Proteínas Nucleares/metabolismo , Proteínas Proto-Oncogénicas/metabolismo , Serina-Treonina Quinasas TOR/metabolismo , Animales , Ciclo Celular , Proteínas de Ciclo Celular , Línea Celular , Proliferación Celular , Supervivencia Celular , Humanos , Masculino , Diana Mecanicista del Complejo 1 de la Rapamicina , Ratones , Neoplasias/genética , Neoplasias/metabolismo , Proteínas Nucleares/genética , Fosforilación , Unión Proteica , Proteínas Proto-Oncogénicas/genética , Proteínas Quinasas S6 Ribosómicas 70-kDa/metabolismo , Transducción de Señal
13.
Stem Cells Int ; 2016: 2683042, 2016.
Artículo en Inglés | MEDLINE | ID: mdl-26880947

RESUMEN

Medulloblastoma (MB) is the most common malignant brain tumor of pediatric age and is characterized by cells expressing stem, astroglial, and neuronal markers. Among them, stem-like cells (hMB-SLCs) represent a fraction of the tumor cell population with the potential of self-renewal and proliferation and have been associated with tumor poor prognosis. In this context, microRNAs have been described as playing a pivotal role in stem cells differentiation. In our paper, we analyze microRNAs profile and genes expression of hMB-SLCs before and after Retinoic Acid- (RA-) induced differentiation. We aimed to identify pivotal players of specific pathways sustaining stemness and/or tumor development and progression and integrate the results of our recent proteomic study. Our results uncovered 22 differentially expressed microRNAs that were used as input together with deregulated genes and proteins in the Genomatix Pathway System (GePS) analysis revealing 3 subnetworks that could be interestingly involved in the maintenance of hMB-SLCs proliferation. Taken together, our findings highlight microRNAs, genes, and proteins that are significantly modulated in hMB-SLCs with respect to their RA-differentiated counterparts and could open new perspectives for prognostic and therapeutic intervention on MB.

14.
Sci Rep ; 6: 18980, 2016 Jan 08.
Artículo en Inglés | MEDLINE | ID: mdl-26742647

RESUMEN

Sex differences play a role in pain sensitivity, efficacy of analgesic drugs and prevalence of neuropathic pain, even if the underlying mechanisms are far from being understood. We demonstrate that male and female mice react differently to structural and functional changes induced by sciatic nerve ligature, used as model of neuropathic pain. Male mice show a gradual decrease of allodynia and a complete recovery while, in females, allodynia and gliosis are still present four months after neuropathy induction. Administration of 17ß-estradiol is able to significantly attenuate this difference, reducing allodynia and inducing a complete recovery also in female mice. Parallel to pain attenuation, 17ß-estradiol treated-mice show a functional improvement of the injured limb, a faster regenerative process of the peripheral nerve and a decreased neuropathy-induced gliosis. These results indicate beneficial effects of 17ß-estradiol on neuropathic pain and neuronal regeneration and focuses on the importance of considering gonadal hormones also in clinical studies.


Asunto(s)
Analgésicos/farmacología , Estradiol/farmacología , Gliosis/tratamiento farmacológico , Hiperalgesia/tratamiento farmacológico , Neuralgia/tratamiento farmacológico , Nervio Ciático/efectos de los fármacos , Animales , Femenino , Perfilación de la Expresión Génica , Regulación de la Expresión Génica , Gliosis/etiología , Gliosis/genética , Gliosis/fisiopatología , Hiperalgesia/etiología , Hiperalgesia/genética , Hiperalgesia/fisiopatología , Queratinas/genética , Queratinas/metabolismo , Ligadura/efectos adversos , Masculino , Ratones , Anotación de Secuencia Molecular , Miosinas/genética , Miosinas/metabolismo , Regeneración Nerviosa/efectos de los fármacos , Regeneración Nerviosa/fisiología , Neuralgia/etiología , Neuralgia/genética , Neuralgia/fisiopatología , Dimensión del Dolor , Umbral del Dolor/efectos de los fármacos , Umbral del Dolor/fisiología , Nervio Ciático/lesiones , Nervio Ciático/metabolismo , Nervio Ciático/fisiopatología , Caracteres Sexuales
15.
Mol Biosyst ; 11(6): 1603-11, 2015 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-25756958

RESUMEN

Human medulloblastoma (MB) is a malignant brain tumor that comprises four distinct molecular subgroups including the Sonic Hedgehog (SHH)-MB group. A leading cause of the SHH subgroup is an aberrant activation of the SHH pathway, a developmental signaling that regulates postnatal development of the cerebellum by promoting the mitotic expansion of granule neural precursors (GNPs) in the external granule layer (EGL). The abnormal SHH signaling pathway drives not only SHH-MB but also its cancer stem-like cells (SLCs), which represent a fraction of the tumor cell population that maintain cancer growth and have been associated with high grade tumors. Here, we report the first proteomic analysis of human SHH-MB SLCs before and after Retinoic Acid (RA)-induced differentiation. A total of 994 nLC-MS buckets were statistically analysed returning 68 modulated proteins between SLCs and their differentiated counterparts. Heat Shock Protein 70 (Hsp70) was one of the proteins that characterized the protein profile of SLCs. By means of Ingenuity Pathway Analysis (IPA), Genomatix analysis and extending the network obtained using the differentially expressed proteins we found a correlation between Hsp70 and the NF-κB complex. A key driver of the SHH-MB group is cMET whose downstream proliferation/survival signalling is indeed via PI3K/Akt/NF-κB. We confirmed the results of the proteomic analysis by western blot, underlining that a P-p65/NF-κB activatory complex is highly expressed in SLCs. Taking together these results we define a new protein feature of SHH-MB SLCs.


Asunto(s)
Proteínas Hedgehog/metabolismo , Meduloblastoma/metabolismo , Células Madre Neoplásicas/metabolismo , Proteoma/análisis , Antineoplásicos/farmacología , Diferenciación Celular/efectos de los fármacos , Humanos , Lactante , Meduloblastoma/tratamiento farmacológico , Meduloblastoma/patología , Células Madre Neoplásicas/efectos de los fármacos , Células Madre Neoplásicas/patología , Proteoma/metabolismo , Proteómica , Transducción de Señal , Tretinoina/farmacología , Células Tumorales Cultivadas
16.
Mol Biosyst ; 10(6): 1332-44, 2014 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-24675778

RESUMEN

Impaired dopamine homeostasis is an early event in the pathogenesis of Parkinson's disease. Generation of intracellular reactive oxygen species consequent to dopamine oxidation leads to mitochondrial dysfunction and eventually cell death. Alterations in the mitochondrial proteome due to dopamine exposure were investigated in the SH-SY5Y human neuroblastoma cell line. The combination of two orthogonal proteomic approaches, two-dimensional electrophoresis and shotgun proteomics (proteomeXchange dataset PXD000838), was used to highlight the specific pathways perturbed by the increase of intracellular dopamine, in comparison with those perturbed by a specific mitochondrial toxin (4-methylphenylpyridinium, MPP(+)), a neurotoxin causing Parkinsonism-like symptoms in animal models. Proteins altered by MPP(+) did not completely overlap with those affected by dopamine treatment. In particular, the MPP(+) target complex I component NADH dehydrogenase [ubiquinone] iron-sulfur protein 3 was not affected by dopamine together with 26 other proteins. The comparison of proteomics approaches highlighted the fragmentation of some mitochondrial proteins, suggesting an alteration of the mitochondrial protease activity. Pathway and disease association analysis of the proteins affected by dopamine revealed the overrepresentation of the Parkinson's disease and the parkin-ubiquitin proteasomal system pathways and of gene ontologies associated with generation of precursor metabolites and energy, response to topologically incorrect proteins and programmed cell death. These alterations may be globally interpreted in part as the result of a direct effect of dopamine on mitochondria (e.g. alteration of the mitochondrial protease activity) and in part as the effect on mitochondria of a general activation of cellular processes (e.g. regulation of programmed cell death).


Asunto(s)
1-Metil-4-fenilpiridinio/farmacología , Dopamina/farmacología , Proteínas Mitocondriales/metabolismo , Neurotoxinas/farmacología , Enfermedad de Parkinson/etiología , Enfermedad de Parkinson/metabolismo , Línea Celular Tumoral , Dopamina/metabolismo , Regulación Neoplásica de la Expresión Génica/efectos de los fármacos , Homeostasis , Humanos , Modelos Neurológicos , Enfermedad de Parkinson/patología , Proteómica , Especies Reactivas de Oxígeno/metabolismo
17.
Mol Biosyst ; 9(6): 1117-26, 2013 Jun.
Artículo en Inglés | MEDLINE | ID: mdl-23609890

RESUMEN

Cystic fibrosis (CF) is an autosomal recessive disorder associated with mutations of the cystic fibrosis transmembrane conductance regulator (CFTR) gene and defective chloride transport across the epithelial cell membranes. Abnormal epithelial ion transport is the primary cause of persistent airway infections and chronic inflammation in CF patients. In order to gain further insight into the mechanisms of epithelial dysfunctions linked to CFTR mutations, we performed and integrated proteomic and ionomic analysis of human bronchial epithelial IB3-1 cells and compared them with a CFTR-complemented isogenic cell line (C38). Aside from changes that were consistent with known effects related to CFTR mutations, such as differences in glycolytic and gluconeogenic pathways and unfolded protein responses, differential proteomics highlighted significant alteration of protein expression and, in particular, of the 14-3-3 signalling pathway that is known to be involved in cellular calcium (Ca) homeostasis. Of note, restoring chloride efflux by acting on Ca cellular homeostasis has been shown to be a promising therapeutic intervention for CF. Ionomic analysis showed significant changes in the IB3-1 element profile compared with C38 cells and in particular we observed an increase of intracellular Ca that significantly correlates with intracellular zinc (Zn) levels, suggesting a synergistic role of Ca and Zn influx. This finding is particularly intriguing because Zn has been reported to be effective in CF treatment increasing Ca influx. Taken together, our proteomic and ionomic data reveal that CFTR mutation sets in motion endogenous mechanisms counteracting impaired chloride transport mainly acting on epithelial ion transport and increasing intracellular Ca, suggesting potential links between protein expression and this response.


Asunto(s)
Calcio/metabolismo , Regulador de Conductancia de Transmembrana de Fibrosis Quística/genética , Regulador de Conductancia de Transmembrana de Fibrosis Quística/metabolismo , Fibrosis Quística/metabolismo , Transporte Iónico/genética , Proteínas 14-3-3/metabolismo , Línea Celular Transformada , Fibrosis Quística/genética , Células Epiteliales/citología , Células Epiteliales/metabolismo , Perfilación de la Expresión Génica , Homeostasis , Humanos , Proteómica , Transducción de Señal/genética , Zinc/metabolismo
18.
Proteomics ; 13(6): 1002-9, 2013 Mar.
Artículo en Inglés | MEDLINE | ID: mdl-23319365

RESUMEN

Transthyretin (TTR) is a homotetrameric protein of the CNS that plays a role of as the major thyroxine (T4) carrier from blood to cerebrospinal fluid (CSF). T4 physiologically helps oligodendrocyte precursor cells to turn into myelinating oligodendrocytes, enhancing remyelination after myelin sheet damage. We investigated post-translational oxidative modifications of serum and CSF TTR in multiple sclerosis subjects, highlighting high levels of S-sulfhydration and S-sulfonation of cysteine in position ten only in the cerebral TTR, which correlate with an anomalous TTR protein folding as well as with disease duration. Moreover, we found low levels of free T4 in CSF of multiple sclerosis patients, suggestive of a potential role of these modifications in T4 transport into the brain.


Asunto(s)
Esclerosis Múltiple/líquido cefalorraquídeo , Prealbúmina/líquido cefalorraquídeo , Procesamiento Proteico-Postraduccional , Adulto , Estudios de Casos y Controles , Femenino , Humanos , Masculino , Persona de Mediana Edad , Oxidación-Reducción , Prealbúmina/química , Prealbúmina/aislamiento & purificación , Isoformas de Proteínas/líquido cefalorraquídeo , Isoformas de Proteínas/química , Isoformas de Proteínas/aislamiento & purificación , Espectrometría de Masa por Láser de Matriz Asistida de Ionización Desorción , Tiroxina/líquido cefalorraquídeo
19.
J Proteomics ; 75(4): 1440-53, 2012 Feb 02.
Artículo en Inglés | MEDLINE | ID: mdl-22146475

RESUMEN

The Ubiquitin-Proteasome System (UPS) and the Autophagy-Lysosome Pathways (ALP) are key mechanisms for cellular homeostasis sustenance and protein clearance. A wide number of Neurodegenerative Diseases (NDs) are tied with UPS impairment and have been also described as proteinopathies caused by aggregate-prone proteins, not efficiently removed by proteasome. Despite the large knowledge on proteasome biological role, molecular mechanisms associated with its impairment are still blur. We have pursued a comprehensive proteomic investigation to evaluate the phenotypic rearrangements in protein repertoires associated with a UPS blockage. Different functional proteomic approaches have been employed to tackle UPS impairment impact on human NeuroBlastoma (NB) cell lines responsive to proteasome inhibition by Epoxomicin. 2-Dimensional Electrophoresis (2-DE) separation combined with Mass Spectrometry and Shotgun Proteomics experiments have been employed to design a thorough picture of protein profile. Unsupervised meta-analysis of the collected proteomic data revealed that all the identified proteins relate each other in a functional network centered on beta-estradiol. Moreover we showed that treatment of cells with beta-estradiol resulted in aggregate removal and increased cell survival due to activation of the autophagic pathway. Our data may provide the molecular basis for the use of beta-estradiol in neurodegenerative disorders by induction of protein aggregate removal.


Asunto(s)
Neoplasias Encefálicas/metabolismo , Estradiol/metabolismo , Neuroblastoma/metabolismo , Complejo de la Endopetidasa Proteasomal/metabolismo , Proteómica/métodos , Ubiquitina/metabolismo , Autofagia , Línea Celular Tumoral , Electroforesis en Gel Bidimensional/métodos , Regulación Neoplásica de la Expresión Génica , Humanos , Lisosomas/metabolismo , Espectrometría de Masas/métodos , Modelos Biológicos , Enfermedades Neurodegenerativas/metabolismo
20.
Clin Cancer Res ; 17(7): 1935-46, 2011 Apr 01.
Artículo en Inglés | MEDLINE | ID: mdl-21307145

RESUMEN

PURPOSE: To determine the in vivo and in vitro antiangiogenic power of lenalidomide, a "lead compound" of IMiD immunomodulatory drugs in bone marrow (BM) endothelial cells (EC) of patients with multiple myeloma (MM) in active phase (MMEC). EXPERIMENTAL DESIGN: The antiangiogenic effect in vivo was studied using the chorioallantoic membrane (CAM) assay. Functional studies in vitro (angiogenesis, "wound" healing and chemotaxis, cell viability, adhesion, and apoptosis) were conducted in both primary MMECs and ECs of patients with monoclonal gammopathies (MGUS) of undetermined significance (MGEC) or healthy human umbilical vein endothelial cells (HUVEC). Real-time reverse transcriptase PCR, Western blotting, and differential proteomic analysis were used to correlate morphologic and biological EC features with the lenalidomide effects at the gene and protein levels. RESULTS: Lenalidomide exerted a relevant antiangiogenic effect in vivo at 1.75 µmol/L, a dose reached in interstitial fluids of patients treated with 25 mg/d. In vitro, lenalidomide inhibited angiogenesis and migration of MMECs, but not of MGECs or control HUVECs, and had no effect on MMEC viability, apoptosis, or fibronectin- and vitronectin-mediated adhesion. Lenalidomide-treated MMECs showed changes in VEGF/VEGFR2 signaling pathway and several proteins controlling EC motility, cytoskeleton remodeling, and energy metabolism pathways. CONCLUSIONS: This study provides information on the molecular mechanisms associated with the antimigratory and antiangiogenic effects of lenalidomide in primary MMECs, thus giving new avenues for effective endothelium-targeted therapies in MM.


Asunto(s)
Inhibidores de la Angiogénesis/farmacología , Células de la Médula Ósea/fisiología , Movimiento Celular/efectos de los fármacos , Células Endoteliales/fisiología , Mieloma Múltiple/tratamiento farmacológico , Talidomida/análogos & derivados , Adulto , Anciano , Inhibidores de la Angiogénesis/uso terapéutico , Animales , Proteínas Reguladoras de la Apoptosis/biosíntesis , Células de la Médula Ósea/efectos de los fármacos , Quimiocina CCL2/biosíntesis , Quimiocina CXCL12/biosíntesis , Pollos , Membrana Corioalantoides/efectos de los fármacos , Membrana Corioalantoides/metabolismo , Medios de Cultivo Condicionados , Células Endoteliales/efectos de los fármacos , Femenino , Factor 2 de Crecimiento de Fibroblastos/biosíntesis , Regulación Neoplásica de la Expresión Génica , Humanos , Péptidos y Proteínas de Señalización Intracelular/metabolismo , Lenalidomida , Masculino , Proteínas de la Membrana/biosíntesis , Persona de Mediana Edad , Mieloma Múltiple/patología , Neovascularización Patológica/tratamiento farmacológico , Proteoma/metabolismo , Proteínas Proto-Oncogénicas/biosíntesis , Selenoproteína W/biosíntesis , Transducción de Señal , Talidomida/farmacología , Talidomida/uso terapéutico , Factor A de Crecimiento Endotelial Vascular/genética , Factor A de Crecimiento Endotelial Vascular/metabolismo , Receptor 2 de Factores de Crecimiento Endotelial Vascular/antagonistas & inhibidores
SELECCIÓN DE REFERENCIAS
DETALLE DE LA BÚSQUEDA